biomarkers for tuberculosis: the case for lipoarabinomannan

13
Biomarkers for tuberculosis: the case for lipoarabinomannan Margarida Correia-Neves 1,2,3 , Gabrielle Fröberg 3,4 , Liudmyla Korshun 5 , Sofia Viegas 6 , Paula Vaz 6,7 , Nehaben Ramanlal 7 , Judith Bruchfeld 3,4 , Beston Hamasur 5,8 , Patrick Brennan 9 and Gunilla Källenius 3 Affiliations: 1 Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal. 2 ICVS/3Bs, PT Government Associate Laboratory, Braga/Guimarães, Portugal. 3 Division of Infectious Diseases, Dept of Medicine Solna, Karolinska Institutet, Stockholm, Sweden. 4 Dept of Infectious Diseases, Karolinska University Hospital, Stockholm, Sweden. 5 Biopromic AB, Solna, Sweden. 6 Instituto Nacional de Saúde, Ministério da Saúde, Maputo, Mozambique. 7 Fundação Ariel Glaser Contra o SIDA Pediátrico, Maputo, Mozambique. 8 Dept of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden. 9 Dept of Microbiology, Immunology and Pathology, Colorado State University, Fort Collins, CO, USA. Correspondence: Gunilla Källenius, Division of Infectious Diseases, Dept of Medicine Solna, Karolinska Institutet, Stockholm 17176, Sweden. E-mail: [email protected] ABSTRACT Tuberculosis (TB) is considered the most onerous of infectious diseases according to recent reports from the World Health Organization. Available tests for TB diagnosis present severe limitations, and a reliable point-of-care (POC) diagnostic test does not exist. Neither is there a test to discern between the different stages of TB, and in particular to predict which patients with Mycobacterium tuberculosis infection and no clinical signs are more at risk of advancing to overt disease. We here review the usefulness of mycobacterial lipoarabinomannan (LAM) as a diagnostic marker for active and latent TB and, also, aspects of the immune response to LAM relevant to such tests. There is a high potential for urinary LAM-based POC tests for the diagnosis of active TB. Some technical challenges to optimised sensitivity of the test will be detailed. A method to quantify LAM in urine or serum should be further explored as a test of treatment effect. Recent data on the immune response to LAM suggest that markers for host response to LAM should be investigated for a prognostic test to recognise individuals at the greatest risk of disease activation. @ERSpublications There is a high potential for a urinary LAM-based point-of-care test to diagnose TB. Markers for host response to LAM should be explored to identify those at highest risk of developing active TB. http://ow.ly/FyCs30n4uFE Cite this article as: Correia-Neves M, Fröberg G, Korshun L, et al. Biomarkers for tuberculosis: the case for lipoarabinomannan. ERJ Open Res 2019; 5: 00115-2018 [https://doi.org/10.1183/ 23120541.00115-2018]. Copyright ©ERS 2019. This article is open access and distributed under the terms of the Creative Commons Attribution Non-Commercial Licence 4.0. Received: July 20 2018 | Accepted after revision: Nov 05 2018 https://doi.org/10.1183/23120541.00115-2018 ERJ Open Res 2019; 5: 00115-2018 REVIEW TUBERCULOSIS

Upload: others

Post on 11-Jan-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Biomarkers for tuberculosis: the case for lipoarabinomannan

Biomarkers for tuberculosis: the case forlipoarabinomannan

Margarida Correia-Neves1,2,3, Gabrielle Fröberg 3,4, Liudmyla Korshun5,Sofia Viegas6, Paula Vaz6,7, Nehaben Ramanlal7, Judith Bruchfeld3,4,Beston Hamasur5,8, Patrick Brennan9 and Gunilla Källenius 3

Affiliations: 1Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho,Braga, Portugal. 2ICVS/3B’s, PT Government Associate Laboratory, Braga/Guimarães, Portugal. 3Division ofInfectious Diseases, Dept of Medicine Solna, Karolinska Institutet, Stockholm, Sweden. 4Dept of InfectiousDiseases, Karolinska University Hospital, Stockholm, Sweden. 5Biopromic AB, Solna, Sweden. 6InstitutoNacional de Saúde, Ministério da Saúde, Maputo, Mozambique. 7Fundação Ariel Glaser Contra o SIDAPediátrico, Maputo, Mozambique. 8Dept of Microbiology, Tumor and Cell Biology, Karolinska Institutet,Stockholm, Sweden. 9Dept of Microbiology, Immunology and Pathology, Colorado State University, FortCollins, CO, USA.

Correspondence: Gunilla Källenius, Division of Infectious Diseases, Dept of Medicine Solna, KarolinskaInstitutet, Stockholm 17176, Sweden. E-mail: [email protected]

ABSTRACT Tuberculosis (TB) is considered the most onerous of infectious diseases according to recentreports from the World Health Organization. Available tests for TB diagnosis present severe limitations,and a reliable point-of-care (POC) diagnostic test does not exist. Neither is there a test to discern betweenthe different stages of TB, and in particular to predict which patients with Mycobacterium tuberculosisinfection and no clinical signs are more at risk of advancing to overt disease. We here review theusefulness of mycobacterial lipoarabinomannan (LAM) as a diagnostic marker for active and latent TBand, also, aspects of the immune response to LAM relevant to such tests. There is a high potential forurinary LAM-based POC tests for the diagnosis of active TB. Some technical challenges to optimisedsensitivity of the test will be detailed. A method to quantify LAM in urine or serum should be furtherexplored as a test of treatment effect. Recent data on the immune response to LAM suggest that markersfor host response to LAM should be investigated for a prognostic test to recognise individuals at thegreatest risk of disease activation.

@ERSpublicationsThere is a high potential for a urinary LAM-based point-of-care test to diagnose TB. Markersfor host response to LAM should be explored to identify those at highest risk of developingactive TB. http://ow.ly/FyCs30n4uFE

Cite this article as: Correia-Neves M, Fröberg G, Korshun L, et al. Biomarkers for tuberculosis: thecase for lipoarabinomannan. ERJ Open Res 2019; 5: 00115-2018 [https://doi.org/10.1183/23120541.00115-2018].

Copyright ©ERS 2019. This article is open access and distributed under the terms of the Creative Commons AttributionNon-Commercial Licence 4.0.

Received: July 20 2018 | Accepted after revision: Nov 05 2018

https://doi.org/10.1183/23120541.00115-2018 ERJ Open Res 2019; 5: 00115-2018

REVIEWTUBERCULOSIS

Page 2: Biomarkers for tuberculosis: the case for lipoarabinomannan

Tuberculosis: a global threat to human healthIt is estimated that about 23% of the global population is infected with Mycobacterium tuberculosis [1] andtuberculosis (TB) accounts for 1.4 million deaths annually and for one-fifth of adult deaths in poor/low-income countries. Each patient with active pulmonary TB, if left untreated, is estimated to infect10–15 other individuals per year [2]. Thus, interrupting disease transmission is of major importance andrequires early detection, in combination with adequate treatment.

Host immune response in TBDepending on the immune response of the host, upon exposure to M. tuberculosis, individuals mighteliminate the bacteria, progress to active TB or develop a persistent infection in the absence of clinicalmanifestation, referred to as latent TB. The initial response is due to the interaction of the unique lipids/glycolipids/carbohydrates/peptidoglycans of the M. tuberculosis cell envelope, with cells of the innateimmune system such as macrophages and dendritic cells [3]. The manner in which macrophages anddendritic cells activate or suppress distinct microbicidal mechanisms, the pattern of cytokine beingproduce and secreted, and how antigens interact with the major histocompatibility complex dictates theprofile of the acquired immune response. The elicited acquired immune response mediated by T-cellsplays a very important part in M. tuberculosis infection control [4]. However, the precise M. tuberculosisantigens and detailed profile of the host immune response necessary for effective acquired immunity toM. tuberculosis have yet to be determined. Most studies of the acquired immune response focus on therole played by antigenic proteins/peptides and very little address the mycobacterial antigens of a lipoglycannature. However, lipoglycan antigens may have been undervalued and might in fact play a crucial part inthe overall immune response to M. tuberculosis and as such be of extreme value for TB diagnosis, a subjectdiscussed further ahead.

While the importance of T-cell immunity is long established, the role of humoral immunity has beenconsidered controversial. There is however increasing recent evidence supporting a role for antibodies andB-cells in the establishment of an effective immune response against M. tuberculosis infection [5–7].During active TB, antibody responses are prominent [8, 9], and antibody levels to particularM. tuberculosis protein antigens may increase before symptoms of active TB [8]. Although people withactive TB have been shown to produce antibodies with a low affinity to M. tuberculosis surface moleculesand with a low ratio of IgG/IgM [10], there is evidence suggesting that specific antibodies might preventM. tuberculosis dissemination. Antibodies in the mucosa may also potentially prevent infection via thisroute [11]. Elevated Ag85A-specific IgG titres have recently been identified as a correlate of a lower risk ofTB disease in the MVA85A vaccine trial [12], indicating a possible role for antibodies in M. tuberculosisprotective immunity. Here also, antibodies specific to mycobacterial glycolipids seem to play a relevantrole [13–15] as discussed in detail later.

The need for better TB diagnostic strategies and novel biomarkersPresently available markers/tests for TB diagnosis exhibit serious limitations, and none is a point-of-care(POC) diagnostic test. There is an intensive search for diagnostic biomarkers for TB [16–18], as well aspredictive markers for progression from latent to active TB [19]. It is increasingly evident that latent TBshould be viewed as part of a continuous spectrum, extending from sterilising immunity, to persistentnonprogressing infection and subclinical infection progressing to active disease [20, 21]. Available tests areunable to distinguish those patients with subclinical progressing infection from those with nonprogressinglatent infection [22–24]. Here we discuss the potential use of LAM in the diagnosis of active TB, inpredicting the outcome following M. tuberculosis infection and in response to treatment.

Recently the World Health Organization (WHO) specified the target product profile (TPP) of the mosturgently needed tests for TB [25]. With the objective of initiating treatment, the specificity should be 98%and for sensitivity the minimum requirements should be an overall sensitivity ⩾65%. Besides the need fora diagnostic test, WHO defined as one of the highest priorities a POC triage test, for systematic screening,able to distinguish individuals who need further confirmatory testing. The TPP for such a test was definedby WHO as overall specificity >70% (optimally >80%) and sensitivity >90% (optimally >95%). To achievethese goals the sensitivity of present tests should be increased, while retaining the high specificity. Inaddition, in line with the WHO goal of TB elimination by 2050, there is a great need for a test thatidentifies patients with latent TB who are on the rise of developing active disease.

Diagnostic laboratory methods for diagnosis of active TB are based on detection of evidence ofM. tuberculosis by acid fast staining or direct culturing of bacilli in sputum specimens. Microscopy lackssensitivity, while mycobacterial culturing, the ultimate diagnostic test for TB, is a prolonged exercise thatmay require up to 6–8 weeks, or even more, to provide results. The GeneXpert MTB/RIF (Cepheid Inc.,

https://doi.org/10.1183/23120541.00115-2018 2

TUBERCULOSIS | M. CORREIA-NEVES ET AL.

Page 3: Biomarkers for tuberculosis: the case for lipoarabinomannan

Sunnyvale, CA, USA), and more recently Xpert Ultra, a superior new generation version of GeneXpert,have become important additions to standard liquid culture.

Diagnosis of the latent forms of TB relies on the tuberculin skin test (TST) or the interferon-γ releaseassays (IGRAs). The TST uses purified tuberculin derivative (PPD), which is nonspecific forM. tuberculosis. The interpretation of a TST needs to take into consideration false positive results inindividuals exposed to ubiquitous non-TB mycobacteria and/or previously vaccinated with BCG. Thecommercial IGRAs (Quantiferon Gold-IT and more recently Quantiferon Plus (QIAGEN, Venlo, theNetherlands) as well as T-spot.TB (Oxford Immunotec, Marlborough, MA, USA) useM. tuberculosis-specific antigens (ESAT-6 and CFP-10) and include test tubes to control for anergy andbackground reactivity. However, neither the TST nor IGRAs can help identifying which of the patientswith latent TB are on the verge of progression to active TB, those in immunological control ofM. tuberculosis nor the ones that exhibit just immunological memory to a previous M. tuberculosisinfection. Thus, although these tests are useful in patient management they have no predictive value forprogression to active TB [22–24, 26, 27].

Ideal diagnostic tests would distinguish between those with actual clinical TB; those with the potentialreactivation/latent form; and those who have been cured. For the usage of the test in all different settings,less intrusive sources of potential biomarkers such as human urine, would be ideal.

An example of a new strategy that could be applied for both latent and active TB lies on thedetermination of particular circulating B lymphocytes in the peripheral blood [28]. It has been shownthat B lymphocytes may discriminate individuals with latent and active TB, namely the ratio ofplasmablast to memory lymphocytes. Interestingly LU et al. [29] reported recently that latent TB isassociated with particular M. tuberculosis-specific antibody Fc functional profiles, and distinct antibodyglycosylation patterns. Antibodies from patients with clinical evidence of TB when compared with thoseconforming to the definition of latent TB, were found to drive phagolysosomal maturation,inflammasome activation, and macrophage killing of intracellular M. tuberculosis [29]. These resultsstrongly suggest a role for Fc-mediated antibody effector functions for an effective immune response toM. tuberculosis [29].

Blood RNA signatures prospectively identified people at risk of developing active TB with a sensitivity of53.7% (42.6–64.3) and a specificity of 82.8% (76.7–86.0) [30]. Interestingly, the same signature was laterfound in four different African countries [31]. This finding strongly validates the potential usefulness ofthe blood RNA signatures as an important tool to identify individuals at higher risk of progression toactive disease.

A different strategy that holds promise is untargeted metabolomic approaches. However, the majority ofmetabolites in human body fluids that could accurately identify patients early in infection have lackedstructural definition [32]; an intriguing exception is the identification of N1-acetylisoputreanine anapparent novel polyamine metabolite in human urine that may have potential as a host-derived biomarkerof M. tuberculosis infection [33].

In a proteomic approach to discover biomarkers for pulmonary TB in urine samples, 10 mycobacterialproteins were identified as unique biomarkers of the active from of TB whereas a different set of 6 proteinswere characteristic of latent TB; partial identifications of these proteins were achieved, notably, isoforms of thePE-PGRS protein family were prominent in both the proteins from patients with active and latent TB [34].

LAM as a biomarker for TB diagnosisLAM, a major lipoglycan (a multiglycosylated extension of the phosphatidylinositol mannoside (PIM)glycolipids) of the mycobacterial cell envelope has been shown to be present in the urine ofM. tuberculosis-infected individuals.

LAM is a major constituent of the M. tuberculosis cell wall/envelope and represents up to 15% of thebacterial mass. It is composed of a mannan “core” (an extension of the PIM structure) which is decoratedby a single branched arabinan chain. In addition, in the case of M. tuberculosis the majority of theterminal arabinose unity of the arabinan chain have attached to them short 2–4 mannose containingoligosaccharides called “mannose capping motifs”; this form of LAM is known as ManLAM [35–37](figure 1). LAM exhibits important and distinct immunomodulatory properties [40, 41]. It is considered avirulence factor associated with the pathogenesis of M. tuberculosis infection. During mycobacterialinfections, LAM is present in body fluids representing a potential biomarker to identify infectedindividuals. In addition, the immune response to LAM might also serve as a diagnostic tool. Theusefulness of LAM as a diagnostic marker will be discussed in the following sections.

https://doi.org/10.1183/23120541.00115-2018 3

TUBERCULOSIS | M. CORREIA-NEVES ET AL.

Page 4: Biomarkers for tuberculosis: the case for lipoarabinomannan

Detection of LAM in body fluids as a method for TB diagnosisTests that detect M. tuberculosis antigens in clinical specimens would potentially offer several advantagesin terms of a specific biomarker of TB [42]. Tests for urinary LAM have generated great enthusiasm andnumerous publications. However, several difficulties have been encountered over the last two decades andthe consensus is that the present test(s) are not sensitive enough to diagnose latent TB nor active TB inHIV-negative patients [43–46]. However, recent results strongly suggest that this is a path that deservesfurther effort. A brief historical perspective will be presented followed by the results obtained with twocommercially available tests and the efforts presently ongoing to render the use of LAM detection in urine,and potentially other body fluids, a useful diagnostic test for TB.

The first reports on the possible use of LAM in urine as a diagnostic test were published in 2001 by ourteam. We showed that LAM is present in urine from individuals with active TB [47–49] as well as miceinjected with sonicated M. tuberculosis [47]. Although we clearly showed that LAM is present in urine ofpatients with active TB, the method used at that time required a laborious concentration step. Thesensitivity achieved by us, as well as by several subsequent groups, did not reach the sensitivity requiredfor a diagnostic test for TB.

Two tests were developed by companies and commercialised but obvious improvements are needed torender this approach of general use for the patients in need. A sandwich ELISA, employing polyclonalantibody preparations, was first marketed by Chemogen Inc. (South Portland, ME, USA) as the“Mtb ELISA” test and was sold as “Clearview TB ELISA” first by Inverness Medical Innovations andlater by Alere Inc (Waltham, MA, USA). A set of clinical studies [50–55] using these commercial testswere performed, resulting in a pooled estimated sensitivity of 14% for patients with active TB and noHIV infection and 51% for patients co-infected with HIV (table 1) [56]. Subsequently a lateral flow test,the Determine TB-LAM Ag (Determine TB-LAM; Alere Inc.), using the same polyclonal antibody

FIGURE 1 Schematic rendition ofthe structure of Mycobacterium spp.LAM. Only the major commonstructural features are shown (seevarious works for more structuraldetail [38, 39]). LAM: lipoarabino-mannan. LAM

D-arabinofuranose

D-mannopyranose

D-myoinositol

Succinyl

Palmitic acid

Tuberculostearic acid

https://doi.org/10.1183/23120541.00115-2018 4

TUBERCULOSIS | M. CORREIA-NEVES ET AL.

Page 5: Biomarkers for tuberculosis: the case for lipoarabinomannan

preparations as in the Clearview ELISA, was produced as a POC test. Although easier to perform, thesensitivity of that test remained disappointingly low, as demonstrated in numerous clinical studies. In asystematic review [57] of 12 studies, 6 evaluated the Determine TB-LAM test to identify patients withactive TB and 6 others evaluated the test for TB screening in HIV-positive adults. It was found thatDetermine TB-LAM has low sensitivity for both diagnosis and screening. The combination of DetermineTB-LAM with sputum microscopy revealed an increased sensitivity for active TB when compared witheither test alone but accompanied by a decreased specificity. Determine TB-LAM was shown to potentiallyimprove the accuracy of active TB diagnosis only for seriously ill patients with HIV-co-infection and lowCD4+ T-cell counts [57].

These tests were most sensitive for patients with profound immunosuppression revealing an inversecorrelation between CD4+ T-cell counts and urine LAM sensitivity [57–59] (figure 2). Pooled sensitivity ofDetermine TB-LAM was 56% (41–70%) and specificity 90% (81–95%) in participants with CD4+ T-cellcounts below or equal to 100 cells·µL−1 [57] with a urinary LAM concentration in the range of ng per mL [60].LAM positivity has been associated with poor prognosis [61] and with increased mortality [44, 62–65] andhas been reported as an independent predictor of mortality [63]. In addition, the usage of bedside LAMtesting as a guide for initiating anti-TB treatment in HIV-infected hospitalised inpatients has beenassociated with decreased mortality [66]. After a long evaluation process, WHO recommended in 2016that the Determine TB-LAM assay might be utilised for TB diagnosis in a small subgroup of patients (i.e.in HIV-infected hospitalised adults with symptoms and signs of TB who either are seriously ill and/orhave CD4+ T-cell counts below or equal to 100 cells·μL−1). Recently, in a randomised controlled trial of2600 HIV-infected hospital inpatients with TB, the addition of urine LAM-based screening using the AlereTB-LAM test, a significant mortality reduction was observed in three subgroups of patients: those with lowCD4+ T-cell counts, severe anaemia or clinically suspected TB [67].

The high, and thus detectable, LAM concentration in urine of patients with advanced immunodeficiencyhas been postulated to depend on a high mycobacterial burden [68] and systemic dissemination ofM. tuberculosis. It seems reasonable that patients with a more confined TB would excrete less LAM.Another interpretation could be that the renal tract is affected in advanced HIV infection with

TABLE 1 Performance of commercial urinary lipoarabinomannan (LAM) tests

Sensitivity % Specificity %

TB-LAM (Clearview) ELISA [56]HIV-positive patients 51 (32–69) 94 (79–98)HIV-negative patients 14 (7–24) 97 (90–99)

Determine (Alere) TB-LAM test [57]HIV-positive patients 45 (29–63) 92 (80–97)HIV-negative patients ND ND

Data are presented as median (range). TB: tuberculosis; ND: not determined.

1.00a)

0.75

0.50

0.25

0.00

Po

ole

d s

en

sit

ivit

y

CD4

<50 51–100 101–200 >200

1.00b)

0.75

0.50

0.25

0.00

Po

ole

d s

pe

cifi

cit

y

CD4

<50 51–100 101–200 >200

FIGURE 2 Correlation between CD4+ T-cell counts and lipoarabinomannan (LAM) detection in urine.Compilation of results from a meta-analysis of 12 different studies of tuberculosis (TB)-LAM for TB diagnosisand screening. Plots of a) sensitivity and b) specificity of TB-LAM stratified by CD4+ T-cell counts. Circlesrepresents the pooled estimates (median), with bars representing 95% credible intervals. Reproduced from[57] with permission from the publisher.

https://doi.org/10.1183/23120541.00115-2018 5

TUBERCULOSIS | M. CORREIA-NEVES ET AL.

Page 6: Biomarkers for tuberculosis: the case for lipoarabinomannan

accompanying mycobacteriuria [69–71]. More studies are required to reach consensus on this topic. Thefinding of LAM in urine prior to the initiation of antiretroviral therapy has also been reported to beassociated with immune reconstitution inflammatory syndrome (IRIS) [60, 72], indicating that LAMdetection in urine may predict the development of TB IRIS. The association of urinary LAM and IRISsupports the idea that high urinary LAM concentration reflects a disseminated disease with high bacterialload. Considering that LAM in urine might, at least in part, reflect the bacterial load in the body,measurement of LAM concentration has been suggested as a tool to monitor the effectiveness of anti-TBtreatment (figure 3) [46, 64, 69].

Patients with latent or incipient TB who are on the verge of developing active TB, hypothetically carrybacteria that are metabolically active and dividing. Accordingly, one expects these individuals to excretesmall amounts of LAM in urine, a hypothesis that needs to be tested with more clinical research. Ifconfirmed, a more sensitive urinary LAM assay could possibly become a test to identify individuals atincreased risk of progressing to active disease.

Detection of LAM in other body fluids has also been investigated. A few efforts have been made to detectLAM in sputum, without much success [58, 74], mainly depending on the viscosity and interfering matrixof the sputum. LAM was rarely detected in cerebrospinal fluids of patients with TB meningitis [75, 76] orin pleural effusions [77]. With respect to extrapulmonary TB, tests for LAM in urine were rarely positivein one study of meningitis cases [78].

Host immune responses to LAM as biomarkers for TBThe source of LAM used in in vitro tests is of utmost importance for the correct dissection of theimmune response to LAM. ManLAM derived from M. tuberculosis strain H37Rv and clinicalM. tuberculosis isolates elicit proinflammatory cytokine responses in human macrophages and dendriticcells [3, 79, 80], while ManLAM from the laboratory strain M. tuberculosis Erdman induces no or just aweak response composed of pro- and anti-inflammatory cytokines [81]. Contamination with, for example,PIMs during the preparation of ManLAM may be the cause of some of the reports showing a weakproinflammatory response for particular ManLAM preparations [79]. Most studies of the adaptiveimmune response against M. tuberculosis have focused on the response to antigenic proteins/peptides andvery little has been done on mycobacterial antigens of carbohydrate nature, in spite of the fact thatmycobacterial glycolipids/lipoglycans are the dominant structural entities of the mycobacterial cell walland have strong and distinct effects on the immune response. Only a few reports show the effect of LAMon the adaptive immune response. Liposomal delivery of LAM was shown to trigger M. tuberculosis-specificT-cell activation [82]; polycytotoxic T-cells from bronchiolar lavage were recently reported to be induced bymycobacterial glycolipids, in particular by LAM, which was associated with protection against M. tuberculosisinfection [83].

Identifying specific markers or cytokine/chemokines of cell activation induced by LAM is an interestingvenue to explore as an alternative to the present IGRA tests, in particular in the differentiation between

234

1

0.1

0.01

Neg.

OD

me

an

ab

ove

cu

t-o

ff

Time since start of anti-TB treatment months

Before ≤1 2–3 >3–8 >8

Median OD for

time-point

FIGURE 3 Signal intensity of Mycobacterium tuberculosis lipoarabinomannan in urine of children withtuberculosis (TB) in a high TB/HIV setting by ELISA at different time points after initiation of anti-TB treatment.Optical density (OD) results for HIV-positive participants are shown in red solid lines, results for HIV-negativeparticipants as black dashed lines. The signal intensity became undetectable after 3 months of treatment insix participants. y-axis shows logarithmic scale for mean OD. Neg.: negative. Reproduced from [73].

https://doi.org/10.1183/23120541.00115-2018 6

TUBERCULOSIS | M. CORREIA-NEVES ET AL.

Page 7: Biomarkers for tuberculosis: the case for lipoarabinomannan

different stages of latent TB as well as a complement for active TB diagnosis. Thus, there is an intensesearch for alternatives to ESAT-6 and CFP-10 as antigens used in the IGRA tests [84], as well asalternative to interferon (IFN)-γ [85–87].

Anti-LAM antibodies as markers of TB diseaseAnti-LAM antibodies are induced both during TB infection [88, 89] and after BCG vaccination [89–91].These antibodies have been associated with M. tuberculosis opsonisation and restriction of intracellulargrowth [90, 91]. Passive transfer of anti-LAM monoclonal antibodies has been shown to confer protectionin mice [13, 14]. Based on these findings a number of commercial tests based on antibody detection weredeveloped [92], but none proved to be specific and sensitive enough. In a policy statement in 2011, WHOrecommended against the usage of these tests while underscoring the relevance of continued investigationof diagnostic strategies based on antibody detection [93]. The majority of these commercial tests weredirected at antibodies specific for protein antigens, except one, the Mycodot test (DynaGen Inc.,Cambridge, MA, USA), that detects antibodies specific for LAM. Studies with the Mycodot test inTanzania, Guinea Bissau and Japan revealed sensitivities and specificities from 16.0% to 67.5% and 84.0%to 97.5%, respectively [94]. Using capsular arabinomannan, presumably devoid of the phosphatidyl inositolanchor and its constituent fatty acids, but otherwise structurally identical, anti-arabinomannan antibodytitres were significantly higher for TB individuals that were HIV-negative than for the ones that were HIVco-infected in one study in Tanzania (p<0.01) [95]. In summary, the few studies performed using theMycodot test indicate a high specificity (in contrast with serological tests for other, protein, antigens), buta low sensitivity. Although people with active TB have been shown to produce antibodies with a lowaffinity to the surface of live M. tuberculosis and with a low ratio of IgG/IgM [10], there is evidencesuggesting that specific antibodies might prevent M. tuberculosis dissemination.

Ongoing efforts to circumvent the technical limitations and render LAM detection inurine a useful test for TB patientsIntensive efforts are presently being made by several research teams to develop a true POC test for TBdiagnosis and patient management based on the simple detection and/or quantification of LAM in urine,with promising progress.

Increasing the diagnostic sensitivity of urinary LAM testsSince the concentration of LAM in urine from HIV-negative adult TB patients is very low, in most cases,the analytical sensitivity of the method used is absolutely crucial. We, and others, have found that the trueconcentration of LAM in urine is in the range of 15 pg·mL−1 to several hundred ng·mL−1 [60, 96–98].Based on our findings that urine concentration renders LAM detectable for most “ordinary” TB patients [47],we developed a simple concentration method. This was based on very efficient monoclonal anti-LAMantibodies in combination with a concentration step using gold coated nanoparticles [97]. With this method,we reached a sensitivity of 82% in HIV-negative patients with active TB. However, some of the componentsof the test were not stable enough, resulting in a shelf life of the test of about 3 months. Second, as shownnext, when the analytical sensitivity increased, the “matrix” effect of the urine became more prominent(i.e. the LAM signal was masked apparently by as yet unidentified components in the urine).

Other approaches to increase the sensitivity and/or the simplicity of the test have been made [99], namely:1) immunoassays exploring single-molecule fluorescence-linked immunosorbent assay [100];2) waveguide-based optical biosensors [101]; or 3) electrochemical detection on a disposable chip [102].Efforts to engineer new anti-LAM antibodies with higher affinity have for instance resulted in recombinantantibodies [103]. None of these approaches have so far reached sufficient sensitivity for clinical use. Morerecently a method applying a copper complex dye within a hydrogel nanocage that captures LAM wasdeveloped, and a sensitivity of 96% and specificity of 81% for diagnosis of pulmonary TB disease inHIV-negative adults was reported [104].

Matrix effect and how to overcome itIn experimental studies we found that urine from TB patients and healthy individuals inhibit LAMdetection at varying degrees, when purified LAM was added to different urine samples. Data shown infigure 4, using a set of 15 urine samples, clearly shows that the matrix effect varies between urine samples,LAM in the spiked urine being detectable for some individuals (see for example samples 1, 2, 9, 100 and11 in figure 4) while for others it is undetectable (see for example samples 3 and 4 in figure 4). Theinhibitory effect (“matrix” effect) varied not only between individuals, but also between samples taken atvarious time points from the same individual (data not shown).

The matrix effect is extremely important for the measurement of urine analytes. The composition of humanurine is highly variable [105] with large variations in concentration of individual components [106, 107],

https://doi.org/10.1183/23120541.00115-2018 7

TUBERCULOSIS | M. CORREIA-NEVES ET AL.

Page 8: Biomarkers for tuberculosis: the case for lipoarabinomannan

containing both organic and inorganic constituents, such as carbohydrates, amino acids, peptides, lipidsand end products of metabolism [108]. These organic entities of the urine matrix, as well as pH andelectrolytes, vary depending on normal physiological variations, health conditions, as well as differences infood consumption and medication intake. In immunoassays the matrix components may mask and/orchange the conformation of either antibodies or LAM epitopes in the assay. This masking effect may varydepending on the antibody and/or of LAM epitopes detected by distinct antibodies as well as the type ofassay used [109]. Reducing the inhibitory effects of patient urines obviously would increase the diagnosticsensitivity and accuracy of a LAM-based urine test.

The matrix effect on the LAM signal is evident also in serum [110]. The matrix effect in the serum hasbeen considered to depend on the faculty of ManLAM to form complexes with antibodies [111] and otherproteins. Using spiked samples, SAKAMURI et al. [112] demonstrated that LAM forms complexes withhigh-density lipoprotein present in the serum. In the case of serum various methods of pre-treatment havebeen tried to overcome the matrix effect using different pre-treatment schemes designed to disruptcomplexing with proteins and other components in serum and shown to improve detection [110, 113].Perchloric acid-based pre-treatment was shown to significantly improve the ability to detect LAM [110].

The importance of pHLow pH most often occurs in malnutrition [114] such as wasting TB disease and might reduce the abilityto detect LAM. We found that adjusting the pH to a neutral level had some positive effect on the LAMsignal. Data depicted in figure 4 reveal that adjusting the pH tends to improve LAM detection and that insome cases is sufficient to render LAM detectable (see for example patients 5, 7, 8 and 12 in figure 4),However, the differences in urinary pH do not exclusively explain the large variations in LAM recoverybetween different urine samples.

Effect of urine concentration on LAM detectionWe, and others, have postulated that the more the urine was concentrated the higher would be the LAMsignal. Indeed, in a recent study, GINA et al. [115] showed that collection of early morning urine (that ismore concentrated) improves the urinary lateral flow LAM assay sensitivity in hospitalised patients with

0.8

1.0

1.2

1.4

0.6

0.4

0.2

0

OD

re

su

lts

Patients

1 2 3 4 5 6 7 8 9 10 11 12 13 14 15 Control

Sample

Sample+LAM

Sample/pH

Sample/pH+LAM

FIGURE 4 Lipoarabinomannan (LAM) signal by ELISA in urine samples from children with suspected tuberculosis. Samples were collected atMachava General Hospital in Maputo, Mozambique, and stored at −80°C. The samples were transported frozen to the laboratory at KarolinskaInstitutet, Sweden. After thawing each urine was divided into four samples: 1) nonmanipulated; 2) spiked with 250 pg·mL−1 LAM; 3) pH adjustedwith 40 mM Tris final concentration; 4) pH adjusted and spiked with LAM. LAM signal was measured by ELISA with absorbance at 450 nmfollowing the protocol described previously [97]. PBS spiked with 250 pg·mL−1 LAM was used as a control. LAM was prepared in house asdescribed previously [79].

https://doi.org/10.1183/23120541.00115-2018 8

TUBERCULOSIS | M. CORREIA-NEVES ET AL.

Page 9: Biomarkers for tuberculosis: the case for lipoarabinomannan

TB and HIV-coinfection. However, we found that the higher the concentration of urinary creatinine,which is a marker of urine concentration, the lower the signal (data not shown), indicating that moreconcentrated urine contains more inhibitory factors. So, it seems that in patients with TB/HIV,co-infection and presumably higher concentrations of LAM in urine, the matrix does not mask the LAMsignal while in HIV-negative patients (presumably with low LAM concentration) the matrix effect isenforced by concentration. We thus tested the hypothesis that diluting urine would reduce the matrixeffect on the LAM signal, a standard method for avoiding the matrix effect in biological assays [107].Indeed, we found that the LAM signal is increased in diluted urine spiked with LAM (data not shown).However, dilution of samples with very low concentration of excreted LAM is ineffective since LAM isdiluted to levels below detection limits.

Effect of sample transportation and storageSince transportation and storage of urine samples is a key issue in clinical studies where testing iscentralised, we investigated the impact of various storage temperatures on the LAM signal in spiked urinesamples. We, and others [101], found that when the samples are stored at room temperature the signaldecreases rapidly, in a matter of hours. Even storage at 4°C causes a significant decrease in the LAMdetection as shown in figure 5 for periods as short as 1, 2 and 4 weeks. These findings are in agreementwith a study showing that after storage of urine for 2 h at room temperature LAM becomes undetectableusing an immunoblot method [101]. We, and others [116], also found that freeze–thawing extensivelyreduces LAM detection.

Actually, the fate of carbohydrate containing molecules may also be affected by chemical reactions in theurine. Thus, the formation of glucose ureide by the interaction of glucose with urea results in ammonia, anevent which may change the pH of urine. Urea in urine can, depending on the time of exposure,temperature and pH, disintegrate into isocyanates that may interact with amines on the antibodies, thuspotentially interfering with the binding of the antibodies with carbohydrates like LAM.

FIGURE 5 Effect of storage of urineat 4°C on lipoarabinomannan (LAM)signal detected by ELISA. Urine wasspiked with various concentrationsof LAM and stored at 4°C. LAMsignal was measured by ELISA withabsorbance at 450 nm following theprotocol described previously [97].

2.0

1.5

1.0

0.5

0

Ab

so

rba

nce

at

45

0 n

m

Time of storage

Day 0

5 ng·mL–1 LAM

2.5 ng·mL–1 LAM

1 ng·mL–1 LAM

0.5 ng·mL–1 LAM

1 week 2 weeks 4 weeks

TABLE 2 Lipoarabinomannan as a potential biomarker for tuberculosis diagnosis

Type of test Present status To be explored

Urine antigen detection Low sensitivity, high specificityUseful in subgroup of HIV-positive

Solve matrix problemsIncrease sensitivity

Generate a true POC testSerum antigen detection Matrix problems

Immune complexesSolve matrix and immune complex

problemsCellular immune response Considered a potential alternative to

present IGRAMarker of protection (polycytotoxic

T-cells)Humoral immune response Low sensitivity, high specificity Usefulness for specific subgroups

Marker of protection

IGRA: interferon-γ release assay; POC: point-of-care.

https://doi.org/10.1183/23120541.00115-2018 9

TUBERCULOSIS | M. CORREIA-NEVES ET AL.

Page 10: Biomarkers for tuberculosis: the case for lipoarabinomannan

Concluding remarksThere are several interesting venues for the use of LAM as a biomarker in the diagnosis of TB (table 2), inparticular the use of a urinary LAM-based POC test. To achieve this goal, a simple concentration step isneeded in combination with a method to circumvent the matrix effect of the urine. A method fordetection, or even quantification, of LAM in serum should be further explored.

If increased sensitivity of an antigen test (urine or serum) for LAM is achieved, LAM should be exploredas a predictive biomarker of the outcomes following M. tuberculosis infection, as well as a marker toevaluate the efficacy of anti-TB therapies.

Markers for host response to LAM (cellular and humoral) should be explored for a possible prognostic testto identify those at highest risk of progressing to active TB.

Conflict of interest: M. Correia-Neves has nothing to disclose. G. Fröberg has nothing to disclose. L. Korshun hasnothing to disclose. S. Viegas has nothing to disclose. P. Vaz reports receiving grants from the Ariel Foundation duringthe conduct of the study. N. Ramanlal reports receiving grants from the Ariel Foundation during the conduct of thestudy. J. Bruchfeld has nothing to disclose. B. Hamasur has nothing to disclose. P. Brennan has nothing to disclose.G. Källenius reports receiving grants from the Optimus Foundation during the conduct of the study and owns shares inTBDiadirect AB.

References1 Houben RM, Dodd PJ. The global burden of latent tuberculosis infection: a re-estimation using mathematical

modelling. PLoS Med 2016; 13: e1002152.2 WHO. Global Tuberculosis Report. 2016.3 Kallenius G, Correia-Neves M, Buteme H, et al. Lipoarabinomannan, and its related glycolipids, induce divergent

and opposing immune responses to Mycobacterium tuberculosis depending on structural diversity andexperimental variations. Tuberculosis (Edinb) 2016; 96: 120–130.

4 Cooper AM. Cell-mediated immune responses in tuberculosis. Annu Rev Immunol 2009; 27: 393–422.5 Achkar JM, Chan J, Casadevall A. B cells and antibodies in the defense against Mycobacterium tuberculosis

infection. Immunol Rev 2015; 264: 167–181.6 Kozakiewicz L, Phuah J, Flynn J, et al. The role of B cells and humoral immunity in Mycobacterium tuberculosis

infection. Adv Exp Med Biol 2013; 783: 225–250.7 Jacobs AJ, Mongkolsapaya J, Screaton GR, et al. Antibodies and tuberculosis. Tuberculosis (Edinb) 2016; 101:

102–113.8 Gennaro ML, Affouf M, Kanaujia GV, et al. Antibody markers of incident tuberculosis among HIV-infected

adults in the USA: a historical prospective study. Int J Tuberc Lung Dis 2007; 11: 624–631.9 Laal S, Samanich KM, Sonnenberg MG, et al. Human humoral responses to antigens of Mycobacterium

tuberculosis: immunodominance of high-molecular-mass antigens. Clin Diagn Lab Immunol 1997; 4: 49–56.10 Perley CC, Frahm M, Click EM, et al. The human antibody response to the surface of Mycobacterium

tuberculosis. PLoS One 2014; 9: e98938.11 Alvarez N, Otero O, Camacho F, et al. Passive administration of purified secretory IgA from human colostrum

induces protection against Mycobacterium tuberculosis in a murine model of progressive pulmonary infection.BMC Immunol 2013; 14: Suppl. 1, S3.

12 Fletcher HA, Snowden MA, Landry B, et al. T-cell activation is an immune correlate of risk in BCG vaccinatedinfants. Nat Commun 2016; 7: 11290.

13 Hamasur B, Haile M, Pawlowski A, et al. A mycobacterial lipoarabinomannan specific monoclonal antibody andits F(ab′) fragment prolong survival of mice infected with Mycobacterium tuberculosis. Clin Exp Immunol 2004;138: 30–38.

14 Teitelbaum R, Glatman-Freedman A, Chen B, et al. A mAb recognizing a surface antigen of Mycobacteriumtuberculosis enhances host survival. Proc Natl Acad Sci USA 1998; 95: 15688–15693.

15 Cardona PJ, Julian E, Valles X, et al. Production of antibodies against glycolipids from the Mycobacteriumtuberculosis cell wall in aerosol murine models of tuberculosis. Scand J Immunol 2002; 55: 639–645.

16 Weiner J, Kaufmann SH. High-throughput and computational approaches for diagnostic and prognostic hosttuberculosis biomarkers. Int J Infect Dis 2017; 56: 258–262.

17 Goletti D, Petruccioli E, Joosten SA, et al. Tuberculosis biomarkers: from diagnosis to protection. Infect Dis Rep2016; 8: 6568.

18 Wallis RS, Doherty TM, Onyebujoh P, et al. Biomarkers for tuberculosis disease activity, cure, and relapse. LancetInfect Dis 2009; 9: 162–172.

19 Petruccioli E, Scriba TJ, Petrone L, et al. Correlates of tuberculosis risk: predictive biomarkers for progression toactive tuberculosis. Eur Respir J 2016; 48: 1751–1763.

20 Barry CE III, Boshoff HI, Dartois V, et al. The spectrum of latent tuberculosis: rethinking the biology andintervention strategies. Nat Rev Microbiol 2009; 7: 845–855.

21 Salgame P, Geadas C, Collins L, et al. Latent tuberculosis infection: revisiting and revising concepts. Tuberculosis(Edinb) 2015; 95: 373–384.

22 Pai M, Denkinger CM, Kik SV, et al. Gamma interferon release assays for detection of Mycobacteriumtuberculosis infection. Clin Microbiol Rev 2014; 27: 3–20.

23 Auguste P, Tsertsvadze A, Pink J, et al. Comparing interferon-gamma release assays with tuberculin skin test foridentifying latent tuberculosis infection that progresses to active tuberculosis: systematic review andmeta-analysis. BMC Infect Dis 2017; 17: 200.

24 Nemes E, Rozot V, Geldenhuys H, et al. Optimization and interpretation of serial QuantiFERON testing tomeasure acquisition of Mycobacterium tuberculosis infection. Am J Respir Crit Care Med 2017; 196: 638–648.

https://doi.org/10.1183/23120541.00115-2018 10

TUBERCULOSIS | M. CORREIA-NEVES ET AL.

Page 11: Biomarkers for tuberculosis: the case for lipoarabinomannan

25 WHO. High-priority target product profiles for new tuberculosis diagnostics. Report of a consensus meeting. 2014.26 Mahomed H, Hawkridge T, Verver S, et al. The tuberculin skin test versus QuantiFERON TB Gold(R) in

predicting tuberculosis disease in an adolescent cohort study in South Africa. PLoS One 2011; 6: e17984.27 Banaei N, Pai M. Detecting new Mycobacterium tuberculosis infection. Time for a more nuanced interpretation

of QuantiFERON conversions. Am J Respir Crit Care Med 2017; 196: 546–547.28 Sebina I, Biraro IA, Dockrell HM, et al. Circulating B-lymphocytes as potential biomarkers of tuberculosis

infection activity. PLoS One 2014; 9: e106796.29 Lu LL, Chung AW, Rosebrock TR, et al. A functional role for antibodies in tuberculosis. Cell 2016; 167: 433–443.30 Zak DE, Penn-Nicholson A, Scriba TJ, et al. A blood RNA signature for tuberculosis disease risk: a prospective

cohort study. Lancet 2016; 387: 2312–2322.31 Suliman S, Thompson E, Sutherland J, et al. Four-gene pan-African blood signature predicts progression to

tuberculosis. Am J Respir Crit Care Med 2018; 197: 1198–1208.32 Mahapatra S, Hess AM, Johnson JL, et al. A metabolic biosignature of early response to anti-tuberculosis

treatment. BMC Infect Dis 2014; 14: 53.33 Fitzgerald BL, Mahapatra S, Farmer DK, et al. Elucidating the structure of N1-acetylisoputreanine: a novel

polyamine catabolite in human urine. ACS Omega 2017; 2: 3921–3930.34 Young BL, Mlamla Z, Gqamana PP, et al. The identification of tuberculosis biomarkers in human urine samples.

Eur Respir J 2014; 43: 1719–1729.35 Chatterjee D, Bozic CM, McNeil M, et al. Structural features of the arabinan component of the

lipoarabinomannan of Mycobacterium tuberculosis. J Biol Chem 1991; 266: 9652–9660.36 Hunter SW, Brennan PJ. Evidence for the presence of a phosphatidylinositol anchor on the lipoarabinomannan

and lipomannan of Mycobacterium tuberculosis. J Biol Chem 1990; 265: 9272–9279.37 Chatterjee D, Bozic C, Aspinall GO, et al. Glucuronic acid- and branched sugar-containing glycolipid antigens of

Mycobacterium avium. J Biol Chem 1988; 263: 4092–4097.38 Kaur D, Angala SK, Wu SW, et al. A single arabinan chain is attached to the phosphatidylinositol mannosyl core

of the major immunomodulatory mycobacterial cell envelope glycoconjugate, lipoarabinomannan. J Biol Chem2014; 289: 30249–30256.

39 Grzegorzewicz AE, de Sousa-d’Auria C, McNeil MR, et al. Assembling of the Mycobacterium tuberculosis cellwall core. J Biol Chem 2016; 291: 18867–18879.

40 Hunter SW, Gaylord H, Brennan PJ. Structure and antigenicity of the phosphorylated lipopolysaccharideantigens from the leprosy and tubercle bacilli. J Biol Chem 1986; 261: 12345–12351.

41 Briken V, Porcelli SA, Besra GS, et al. Mycobacterial lipoarabinomannan and related lipoglycans: from biogenesisto modulation of the immune response. Mol Microbiol 2004; 53: 391–403.

42 Flores LL, Steingart KR, Dendukuri N, et al. Systematic review and meta-analysis of antigen detection tests forthe diagnosis of tuberculosis. Clin Vaccine Immunol 2011; 18: 1616–1627.

43 Sahle SN, Asress DT, Tullu KD, et al. Performance of point-of-care urine test in diagnosing tuberculosis suspectswith and without HIV infection in selected peripheral health settings of Addis Ababa, Ethiopia. BMC Res Notes2017; 10: 74.

44 Suwanpimolkul G, Kawkitinarong K, Manosuthi W, et al. Utility of urine lipoarabinomannan (LAM) indiagnosing tuberculosis and predicting mortality with and without HIV: prospective TB cohort from theThailand Big City TB Research Network. Int J Infect Dis 2017; 59: 96–102.

45 Nicol MP, Allen V, Workman L, et al. Urine lipoarabinomannan testing for diagnosis of pulmonary tuberculosisin children: a prospective study. Lancet Glob Health 2014; 2: e278–e284.

46 Kroidl I, Clowes P, Reither K, et al. Performance of urine lipoarabinomannan assays for paediatric tuberculosis inTanzania. Eur Respir J 2015; 46: 761–770.

47 Hamasur B, Bruchfeld J, Haile M, et al. Rapid diagnosis of tuberculosis by detection of mycobacteriallipoarabinomannan in urine. J Microbiol Methods 2001; 45: 41–52.

48 Tessema TA, Hamasur B, Bjun G, et al. Diagnostic evaluation of urinary lipoarabinomannan at an Ethiopiantuberculosis centre. Scand J Infect Dis 2001; 33: 279–284.

49 Tessema TA, Bjune G, Assefa G, et al. Clinical and radiological features in relation to urinary excretion oflipoarabinomannan in Ethiopian tuberculosis patients. Scand J Infect Dis 2002; 34: 167–171.

50 Boehme C, Molokova E, Minja F, et al. Detection of mycobacterial lipoarabinomannan with an antigen-captureELISA in unprocessed urine of Tanzanian patients with suspected tuberculosis. Trans R Soc Trop Med Hyg 2005;99: 893–900.

51 Mutetwa R, Boehme C, Dimairo M, et al. Diagnostic accuracy of commercial urinary lipoarabinomannandetection in African tuberculosis suspects and patients. Int J Tuberc Lung Dis 2009; 13: 1253–1259.

52 Reither K, Saathoff E, Jung J, et al. Low sensitivity of a urine LAM-ELISA in the diagnosis of pulmonarytuberculosis. BMC Infect Dis 2009; 9: 141.

53 Shah M, Variava E, Holmes CB, et al. Diagnostic accuracy of a urine lipoarabinomannan test for tuberculosis inhospitalized patients in a High HIV prevalence setting. J Acquir Immune Defic Syndr 2009; 52: 145–151.

54 Shah M, Martinson NA, Chaisson RE, et al. Quantitative analysis of a urine-based assay for detection oflipoarabinomannan in patients with tuberculosis. J Clin Microbiol 2010; 48: 2972–2974.

55 Gounder CR, Kufa T, Wada NI, et al. Diagnostic accuracy of a urine lipoarabinomannan enzyme-linkedimmunosorbent assay for screening ambulatory HIV-infected persons for tuberculosis. J Acquir Immune DeficSyndr 2011; 58: 219–223.

56 Minion J, Leung E, Talbot E, et al. Diagnosing tuberculosis with urine lipoarabinomannan: systematic review andmeta-analysis. Eur Respir J 2011; 38: 1398–1405.

57 Shah M, Hanrahan C, Wang ZY, et al. Lateral flow urine lipoarabinomannan assay for detecting activetuberculosis in HIV-positive adults. Cochrane Database Syst Rev 2016; 5: CD011420.

58 Dheda K, Davids V, Lenders L, et al. Clinical utility of a commercial LAM-ELISA assay for TB diagnosis inHIV-infected patients using urine and sputum samples. PLoS One 2010; 5: e9848.

59 Swaminathan S, Rekha VV. Antigen detection as a point-of-care test for TB: the case of lipoarabinomannan.Future Microbiol 2012; 7: 559–564.

https://doi.org/10.1183/23120541.00115-2018 11

TUBERCULOSIS | M. CORREIA-NEVES ET AL.

Page 12: Biomarkers for tuberculosis: the case for lipoarabinomannan

60 Conesa-Botella A, Loembe MM, Manabe YC, et al. Urinary lipoarabinomannan as predictor for the tuberculosisimmune reconstitution inflammatory syndrome. J Acquir Immune Defic Syndr 2011; 58: 463–468.

61 Lawn SD, Kerkhoff AD, Burton R, et al. Diagnostic accuracy, incremental yield and prognostic value ofDetermine TB-LAM for routine diagnostic testing for tuberculosis in HIV-infected patients requiring acutehospital admission in South Africa: a prospective cohort. BMC Med 2017; 15: 67.

62 Balcha TT, Winqvist N, Sturegard E, et al. Detection of lipoarabinomannan in urine for identification ofactive tuberculosis among HIV-positive adults in Ethiopian health centres. Trop Med Int Health 2014; 19:734–742.

63 Gupta-Wright A, Peters JA, Flach C, et al. Detection of lipoarabinomannan (LAM) in urine is an independentpredictor of mortality risk in patients receiving treatment for HIV-associated tuberculosis in sub-Saharan Africa:a systematic review and meta-analysis. BMC Med 2016; 14: 53.

64 Drain PK, Gounder L, Grobler A, et al. Urine lipoarabinomannan to monitor anti-tuberculosis therapy responseand predict mortality in an HIV-endemic region: a prospective cohort study. BMJ Open 2015; 5: e006833.

65 Manabe YC, Nonyane BA, Nakiyingi L, et al. Point-of-care lateral flow assays for tuberculosis and cryptococcalantigenuria predict death in HIV infected adults in Uganda. PLoS One 2014; 9: e101459.

66 Peter JG, Zijenah LS, Chanda D, et al. Effect on mortality of point-of-care, urine-based lipoarabinomannantesting to guide tuberculosis treatment initiation in HIV-positive hospital inpatients: a pragmatic, parallel-group,multicountry, open-label, randomised controlled trial. Lancet 2016; 387: 1187–1197.

67 Gupta-Wright A, Corbett EL, van Oosterhout JJ, et al. Rapid urine-based screening for tuberculosis inHIV-positive patients admitted to hospital in Africa (STAMP): a pragmatic, multicentre, parallel-group,double-blind, randomised controlled trial. Lancet 2018; 392: 292–301.

68 Nakiyingi L, Ssengooba W, Nakanjako D, et al. Predictors and outcomes of mycobacteremia amongHIV-infected smear- negative presumptive tuberculosis patients in Uganda. BMC Infect Dis 2015; 15: 62.

69 Wood R, Racow K, Bekker LG, et al. Lipoarabinomannan in urine during tuberculosis treatment: association withhost and pathogen factors and mycobacteriuria. BMC Infect Dis 2012; 12: 47.

70 Cox JA, Lukande RL, Kalungi S, et al. Is urinary lipoarabinomannan the result of renal tuberculosis? Assessmentof the renal histology in an autopsy cohort of Ugandan HIV-Infected adults. PLoS One 2015; 10: e0123323.

71 Lawn SD, Gupta-Wright A. Detection of lipoarabinomannan (LAM) in urine is indicative of disseminated TBwith renal involvement in patients living with HIV and advanced immunodeficiency: evidence and implications.Trans R Soc Trop Med Hyg 2016; 110: 180–185.

72 Lawn SD, Edwards DJ, Kranzer K, et al. Urine lipoarabinomannan assay for tuberculosis screening beforeantiretroviral therapy diagnostic yield and association with immune reconstitution disease. AIDS 2009; 23:1875–1880.

73 Kroidl I, Clowes P, Reither K, et al. Performance of urine lipoarabinomannan assays for paediatric tuberculosis inTanzania. Eur Respir J 2015; 46: 761–770.

74 Peter JG, Cashmore TJ, Meldau R, et al. Diagnostic accuracy of induced sputum LAM ELISA for tuberculosisdiagnosis in sputum-scarce patients. Int J Tuberc Lung Dis 2012; 16: 1108–1112.

75 Patel VB, Bhigjee AI, Paruk HF, et al. Utility of a novel lipoarabinomannan assay for the diagnosis oftuberculous meningitis in a resource-poor high-HIV prevalence setting. Cerebrospinal Fluid Res 2009; 6: 13.

76 Bahr NC, Tugume L, Boulware DR. A word of caution in considering the use of the lipoarabinomannan lateralflow assay on cerebrospinal fluid for detection of tuberculous meningitis. J Clin Microbiol 2016; 54: 241–242.

77 Dheda K, Van-Zyl Smit RN, Sechi LA, et al. Clinical diagnostic utility of IP-10 and LAM antigen levels for thediagnosis of tuberculous pleural effusions in a high burden setting. PLoS One 2009; 4: e4689.

78 Blok N, Visser DH, Solomons R, et al. Lipoarabinomannan enzyme-linked immunosorbent assay for earlydiagnosis of childhood tuberculous meningitis. Int J Tuberc Lung Dis 2014; 18: 205–210.

79 Mazurek J, Ignatowicz L, Kallenius G, et al. Divergent effects of mycobacterial cell wall glycolipids on maturationand function of human monocyte-derived dendritic cells. PLoS One 2012; 7: e42515.

80 Yonekawa A, Saijo S, Hoshino Y, et al. Dectin-2 is a direct receptor for mannose-capped lipoarabinomannan ofmycobacteria. Immunity 2014; 41: 402–413.

81 Bradbury MG, Moreno C. Effect of lipoarabinomannan and mycobacteria on tumour necrosis factor productionby different populations of murine macrophages. Clin Exp Immunol 1993; 94: 57–63.

82 Kallert S, Zenk SF, Walther P, et al. Liposomal delivery of lipoarabinomannan triggers Mycobacteriumtuberculosis specific T-cells. Tuberculosis (Edinb) 2015; 95: 452–462.

83 Busch M, Herzmann C, Kallert S, et al. Lipoarabinomannan-responsive polycytotoxic t cells are associated withprotection in human tuberculosis. Am J Respir Crit Care Med 2016; 194: 345–355.

84 Chegou NN, Black GF, Kidd M, et al. Host markers in QuantiFERON supernatants differentiate active TB fromlatent TB infection: preliminary report. BMC Pulm Med 2009; 9: 21.

85 Frahm M, Goswami ND, Owzar K, et al. Discriminating between latent and active tuberculosis with multiplebiomarker responses. Tuberculosis (Edinb) 2011; 91: 250–256.

86 Jeong YH, Hur YG, Lee H, et al. Discrimination between active and latent tuberculosis based on ratio ofantigen-specific to mitogen-induced IP-10 production. J Clin Microbiol 2015; 53: 504–510.

87 Nonghanphithak D, Reechaipichitkul W, Namwat W, et al. Chemokines additional to IFN-gamma can be usedto differentiate among Mycobacterium tuberculosis infection possibilities and provide evidence of an earlyclearance phenotype. Tuberculosis (Edinb) 2017; 105: 28–34.

88 Tessema TA, Bjune G, Hamasur B, et al. Circulating antibodies to lipoarabinomannan in relation to sputummicroscopy, clinical features and urinary anti-lipoarabinomannan detection in pulmonary tuberculosis. Scand JInfect Dis 2002; 34: 97–103.

89 Brown RM, Cruz O, Brennan M, et al. Lipoarabinomannan-reactive human secretory immunoglobulin Aresponses induced by mucosal bacille Calmette–Guerin vaccination. J Infect Dis 2003; 187: 513–517.

90 de Valliere S, Abate G, Blazevic A, et al. Enhancement of innate and cell-mediated immunity byantimycobacterial antibodies. Infect Immun 2005; 73: 6711–6720.

91 Chen T, Blanc C, Eder AZ, et al. Association of human antibodies to arabinomannan with enhancedmycobacterial opsonophagocytosis and intracellular growth reduction. J Infect Dis 2016; 214: 300–310.

https://doi.org/10.1183/23120541.00115-2018 12

TUBERCULOSIS | M. CORREIA-NEVES ET AL.

Page 13: Biomarkers for tuberculosis: the case for lipoarabinomannan

92 Steingart KR, Flores LL, Dendukuri N, et al. Commercial serological tests for the diagnosis of active pulmonaryand extrapulmonary tuberculosis: an updated systematic review and meta-analysis. PLoS Med 2011; 8: e1001062.

93 WHO. Commercial sero-diagnostic tests for diagnosis of tuberculosis: policy statement. 2011.94 Somi GR, O’Brien RJ, Mfinanga GS, et al. Evaluation of the MycoDot test in patients with suspected tuberculosis

in a field setting in Tanzania. Int J Tuberc Lung Dis 1999; 3: 231–238.95 Yu X, Prados-Rosales R, Jenny-Avital ER, et al. Comparative evaluation of profiles of antibodies to mycobacterial

capsular polysaccharides in tuberculosis patients and controls stratified by HIV status. Clin Vaccine Immunol2012; 19: 198–208.

96 Savolainen L, Kantele A, Sandboge B, et al. Modification of Clearview tuberculosis (TB) enzyme-linkedimmunosorbent assay for TB patients not infected with HIV. Clin Vaccine Immunol 2013; 20: 1479–1482.

97 Hamasur B, Bruchfeld J, van Helden P, et al. A sensitive urinary lipoarabinomannan test for tuberculosis. PLoSOne 2015; 10: e0123457.

98 De P, Amin AG, Valli E, et al. Estimation of D-arabinose by gas chromatography/mass spectrometry as surrogatefor mycobacterial lipoarabinomannan in human urine. PLoS One 2015; 10: e0144088.

99 Sarkar S, Tang XL, Das D, et al. A bispecific antibody based assay shows potential for detecting tuberculosis inresource constrained laboratory settings. PLoS One 2012; 7: e32340.

100 Schmidt R, Jacak J, Schirwitz C, et al. Single-molecule detection on a protein-array assay platform for theexposure of a tuberculosis antigen. J Proteome Res 2011; 10: 1316–1322.

101 Mukundan H, Kumar S, Price DN, et al. Rapid detection of Mycobacterium tuberculosis biomarkers in asandwich immunoassay format using a waveguide-based optical biosensor. Tuberculosis (Edinb) 2012; 92:407–416.

102 Wang L, Leng C, Tang S, et al. Enzyme-free signal amplification for electrochemical detection of Mycobacteriumlipoarabinomannan antibody on a disposable chip. Biosens Bioelectron 2012; 38: 421–424.

103 Chan CE, Gotze S, Seah GT, et al. The diagnostic targeting of a carbohydrate virulence factor fromM. tuberculosis. Sci Rep 2015; 5: 10281.

104 Paris L, Magni R, Zaidi F, et al. Urine lipoarabinomannan glycan in HIV-negative patients with pulmonarytuberculosis correlates with disease severity. Sci Transl Med 2017; 9: eaal2807.

105 Sviridov D, Hortin GL. Urine albumin measurement: effects of urine matrix constituents. Clin Chim Acta 2009;404: 140–143.

106 Simerville JA, Maxted WC, Pahira JJ. Urinalysis: a comprehensive review. Am Fam Physician 2005; 71:1153–1162.

107 Taylor TP, Janech MG, Slate EH, et al. Overcoming the effects of matrix interference in the measurement ofurine protein analytes. Biomark Insights 2012; 7: 1–8.

108 Bouatra S, Aziat F, Mandal R, et al. The human urine metabolome. PLoS One 2013; 8: e73076.109 Boscato LM, Stuart MC. Heterophilic antibodies: a problem for all immunoassays. Clin Chem 1988; 34: 27–33.110 Laurentius LB, Crawford AC, Mulvihill TS, et al. Importance of specimen pretreatment for the low-level

detection of mycobacterial lipoarabinomannan in human serum. Analyst 2016; 142: 177–185.111 Bhattacharya A, Ranadive SN, Kale M, et al. Antibody-based enzyme-linked immunosorbent assay for

determination of immune complexes in clinical tuberculosis. Am Rev Respir Dis 1986; 134: 205–209.112 Sakamuri RM, Price DN, Lee M, et al. Association of lipoarabinomannan with high density lipoprotein in blood:

implications for diagnostics. Tuberculosis (Edinb) 2013; 93: 301–307.113 Crawford AC, Laurentius LB, Mulvihill TS, et al. Detection of the tuberculosis antigenic marker mannose-capped

lipoarabinomannan in pretreated serum by surface-enhanced Raman scattering. Analyst 2016; 142: 186–196.114 Klahr S, Tripathy K, Lotero H. Renal regulation of acid-base balance in malnourished man. Am J Med 1970; 48:

325–331.115 Gina P, Randall PJ, Muchinga TE, et al. Early morning urine collection to improve urinary lateral flow LAM

assay sensitivity in hospitalised patients with HIV-TB co-infection. BMC Infect Dis 2017; 17: 339.116 Lawn SD, Dheda K, Kerkhoff AD, et al. Determine TB-LAM lateral flow urine antigen assay for HIV-associated

tuberculosis: recommendations on the design and reporting of clinical studies. BMC Infect Dis 2013; 13: 407.

https://doi.org/10.1183/23120541.00115-2018 13

TUBERCULOSIS | M. CORREIA-NEVES ET AL.